101
|
Zhou L, Ding Y, Gao Y, Yang B, Bao J, Ma J. Genetic influence on bleeding and over-anticoagulation risk in patients undergoing warfarin treatment after heart valve replacements. Expert Opin Drug Metab Toxicol 2020; 16:1-9. [DOI: 10.1080/17425255.2020.1711883] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Ling Zhou
- Department of Pharmacy, Soochow University, Suzhou, China
| | - Yinglong Ding
- Department of Cardiovascular Surgery, Soochow University, Suzhou, China
| | - Yuan Gao
- Department of Pharmacy, Soochow University, Suzhou, China
| | - Biwen Yang
- Department of Cardiovascular Surgery, Soochow University, Suzhou, China
| | - Jianan Bao
- Department of Pharmacy, Soochow University, Suzhou, China
| | - Jingjing Ma
- Department of Pharmacy, Soochow University, Suzhou, China
| |
Collapse
|
102
|
Elewa H, Awaisu A. Pharmacogenomics In Pharmacy Practice: Current Perspectives. INTEGRATED PHARMACY RESEARCH AND PRACTICE 2019; 8:97-104. [PMID: 31807435 PMCID: PMC6850702 DOI: 10.2147/iprp.s180154] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 11/01/2019] [Indexed: 01/07/2023] Open
Abstract
Pharmacogenomics (i.e., the application of genetic information in predicting an individual's response to drug therapy) plays an increasingly important role in drug development and decision-making regarding precision medicine. This has been shown to reduce the risk of adverse events and improve patient health-care outcomes through targeted therapies and dosing. As the field of pharmacogenomics rapidly evolves, the role of pharmacists in the education, implementation, and research applications of pharmacogenomics is becoming increasingly recognized. This paper aims to provide an overview and current perspectives of pharmacogenomics in contemporary clinical pharmacy practice and to discuss the future directions on advancing pharmacogenomics education, application, and research in pharmacy practice.
Collapse
Affiliation(s)
- Hazem Elewa
- College of Pharmacy, Qatar University Health, Qatar University, Doha, Qatar
| | - Ahmed Awaisu
- College of Pharmacy, Qatar University Health, Qatar University, Doha, Qatar
| |
Collapse
|
103
|
Ragia G, Manolopoulos VG. Pharmacogenomics of anticoagulation therapy: the last 10 years. Pharmacogenomics 2019; 20:1113-1117. [DOI: 10.2217/pgs-2019-0149] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Affiliation(s)
- Georgia Ragia
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
- DNALEX SA, Leontaridou 2, Alexandroupolis, Greece
| | - Vangelis G Manolopoulos
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
- Clinical Pharmacology & Pharmacogenetics Unit, Academic General Hospital of Alexandroupolis, Alexandroupolis, Greece
| |
Collapse
|
104
|
|
105
|
Thorn CF, Whirl-Carrillo M, Hachad H, Johnson JA, McDonagh EM, Ratain MJ, Relling MV, Scott SA, Altman RB, Klein TE. Essential Characteristics of Pharmacogenomics Study Publications. Clin Pharmacol Ther 2019; 105:86-91. [PMID: 30406943 DOI: 10.1002/cpt.1279] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 11/02/2018] [Indexed: 12/17/2022]
Abstract
Pharmacogenomics (PGx) can be seen as a model for biomedical studies: it includes all disease areas of interest and spans in vitro studies to clinical trials, while focusing on the relationships between genes and drugs and the resulting phenotypes. This review will examine different characteristics of PGx study publications and provide examples of excellence in framing PGx questions and reporting their resulting data in a way that maximizes the knowledge that can be built on them.
Collapse
Affiliation(s)
- Caroline F Thorn
- Department of Biomedical Data Sciences, Stanford University, Stanford, California, USA
| | | | - Houda Hachad
- Translational Software, Bellevue, Washington, USA
| | - Julie A Johnson
- College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | | | - Mark J Ratain
- Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Mary V Relling
- Pharmaceutical Department, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Stuart A Scott
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Sema4, a Mount Sinai Venture, Stamford, Connecticut, USA
| | - Russ B Altman
- Department of Genetics, Department of Computer Science, Department of Biomedical Engineering, Stanford University, Stanford, California, USA.,Department of Medicine, Stanford University, Stanford, California, USA
| | - Teri E Klein
- Department of Biomedical Data Sciences, Stanford University, Stanford, California, USA
| |
Collapse
|
106
|
Gardner T, Vazquez SR, Kim K, Jones AE, Witt DM. Providers' utilization and perceptions of warfarin dosing algorithms. Thromb Res 2019; 183:4-12. [PMID: 31505378 DOI: 10.1016/j.thromres.2019.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 07/15/2019] [Accepted: 09/02/2019] [Indexed: 10/26/2022]
Abstract
INTRODUCTION Warfarin dosing algorithms have proven beneficial in increasing time within therapeutic range (TTR) and decreasing adverse events associated with out-of-range international normalized ratios (INRs). Despite widespread availability, providers' utilization and perceptions of warfarin algorithms in real-world practice are unclear. Identifying perceptions and barriers to algorithm use may help attempts to improve warfarin therapy management. METHODS Anticoagulation providers' utilization and perceptions of warfarin dosing algorithms were assessed via a nationwide electronic survey. RESULTS Of the 246 providers who completed the survey, 82% were pharmacists, and 69% had over five years' experience dosing warfarin. Warfarin dosing algorithms were deemed beneficial by 84% of respondents and 72% currently use a warfarin dosing algorithm in their practice at least occasionally. Pharmacists were least likely of anticoagulation providers to use algorithms, although this was not statistically significant (p = 0.12). Algorithm utilization also decreased as years of warfarin dosing experience increased, with the highest rate of usage noted in the first year of dosing warfarin. The most common reason providers gave for discontinuing algorithm use was that they no longer felt it was needed. In this study, clinic patient volume did not appear to be associated with algorithm utilization. CONCLUSION Warfarin dosing algorithms are frequently used among anticoagulation providers, especially those new to dosing warfarin, but use is frequently not sustained over the long-term. Education on the continued benefits of warfarin dosing algorithms could increase long-term utilization, potentially improving patient outcomes.
Collapse
Affiliation(s)
- Trevor Gardner
- University of Utah College of Pharmacy, Department of Pharmacotherapy, University of Utah, 30 South 2000 East Salt, Lake City, UT 84112, United States of America
| | - Sara R Vazquez
- University of Utah Health Thrombosis Center, 50 N Medical Drive Room 1R211, Salt Lake City, UT 84132, United States of America.
| | - Kibum Kim
- University of Utah College of Pharmacy, Department of Pharmacotherapy, University of Utah, 30 South 2000 East Salt, Lake City, UT 84112, United States of America
| | - Aubrey E Jones
- University of Utah College of Pharmacy, Department of Pharmacotherapy, University of Utah, 30 South 2000 East Salt, Lake City, UT 84112, United States of America; University of Utah Health Thrombosis Center, 50 N Medical Drive Room 1R211, Salt Lake City, UT 84132, United States of America
| | - Daniel M Witt
- University of Utah College of Pharmacy, Department of Pharmacotherapy, University of Utah, 30 South 2000 East Salt, Lake City, UT 84112, United States of America; University of Utah Health Thrombosis Center, 50 N Medical Drive Room 1R211, Salt Lake City, UT 84132, United States of America
| |
Collapse
|
107
|
Affiliation(s)
- Stephanie Ross
- From the Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Ontario, Canada (S.R., G.P.).,Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada (G.P.).,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada (G.P.).,Thrombosis and Atherosclerosis Research Institute, Hamilton Health Sciences and McMaster University, Canada (G.P.)
| | - Guillaume Paré
- From the Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Ontario, Canada (S.R., G.P.).,Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada (G.P.).,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada (G.P.).,Thrombosis and Atherosclerosis Research Institute, Hamilton Health Sciences and McMaster University, Canada (G.P.)
| |
Collapse
|
108
|
Roden DM, Van Driest SL, Wells QS, Mosley JD, Denny JC, Peterson JF. Opportunities and Challenges in Cardiovascular Pharmacogenomics: From Discovery to Implementation. Circ Res 2019; 122:1176-1190. [PMID: 29700066 DOI: 10.1161/circresaha.117.310965] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
This review will provide an overview of the principles of pharmacogenomics from basic discovery to implementation, encompassing application of tools of contemporary genome science to the field (including areas of apparent divergence from disease-based genomics), a summary of lessons learned from the extensively studied drugs clopidogrel and warfarin, the current status of implementing pharmacogenetic testing in practice, the role of genomics and related tools in the drug development process, and a summary of future opportunities and challenges.
Collapse
Affiliation(s)
- Dan M Roden
- From the Department of Medicine (D.M.R., S.L.V.D., Q.S.W., J.D.M., J.C.D., J.F.P.) .,Department of Pharmacology (D.M.R., Q.S.W.).,Department of Biomedical Informatics (D.M.R., J.C.D., J.F.P.)
| | - Sara L Van Driest
- From the Department of Medicine (D.M.R., S.L.V.D., Q.S.W., J.D.M., J.C.D., J.F.P.).,Department of Pediatrics (S.L.V.D.), Vanderbilt University Medical Center, Nashville, TN
| | - Quinn S Wells
- From the Department of Medicine (D.M.R., S.L.V.D., Q.S.W., J.D.M., J.C.D., J.F.P.).,Department of Pharmacology (D.M.R., Q.S.W.)
| | - Jonathan D Mosley
- From the Department of Medicine (D.M.R., S.L.V.D., Q.S.W., J.D.M., J.C.D., J.F.P.)
| | - Joshua C Denny
- From the Department of Medicine (D.M.R., S.L.V.D., Q.S.W., J.D.M., J.C.D., J.F.P.).,Department of Biomedical Informatics (D.M.R., J.C.D., J.F.P.)
| | - Josh F Peterson
- From the Department of Medicine (D.M.R., S.L.V.D., Q.S.W., J.D.M., J.C.D., J.F.P.).,Department of Biomedical Informatics (D.M.R., J.C.D., J.F.P.)
| |
Collapse
|
109
|
Abstract
Warfarin is a widely used anticoagulant with a narrow therapeutic index and large interpatient variability in the therapeutic dose. Complications from inappropriate warfarin dosing are one of the most common reasons for emergency room visits. Approximately one third of warfarin dose variability results from common genetic variants. Therefore, it is very necessary to recognize warfarin sensitivity in individuals caused by genetic variants. Based on combined polymorphisms in CYP2C9 and VKORC1, we established a clinical classification for warfarin sensitivity. In the International Warfarin Pharmacogenetic Consortium (IWPC) with 5542 patients, we found that 95.1% of the Black in the IWPC cohort were normal warfarin responders, while 74.8% of the Asian were warfarin sensitive (P < 0.001). Moreover, we created a clinical algorithm to predict warfarin sensitivity in individual patients using logistic regression. Compared to a fixed-dose approach, the clinical algorithm provided significantly better performance. In addition, we validated the derived clinical algorithm using the external Easton cohort with 106 chronic warfarin users. The AUC was 0.836 vs. 0.867 for the Easton cohort and the IWPC cohort, respectively. With the use of this algorithm, it is very likely to facilitate patient care regarding warfarin therapy, thereby improving clinical outcomes.
Collapse
|
110
|
Gage BF, Bass AR, Lin H, Woller SC, Stevens SM, Al-Hammadi N, Anderson JL, Li J, Rodriguez T, Miller JP, McMillin GA, Pendleton RC, Jaffer AK, King CR, Whipple B, Porche-Sorbet R, Napoli L, Merritt K, Thompson AM, Hyun G, Hollomon W, Barrack RL, Nunley RM, Moskowitz G, Dávila-Román V, Eby CS. Effect of Low-Intensity vs Standard-Intensity Warfarin Prophylaxis on Venous Thromboembolism or Death Among Patients Undergoing Hip or Knee Arthroplasty: A Randomized Clinical Trial. JAMA 2019; 322:834-842. [PMID: 31479138 PMCID: PMC6724181 DOI: 10.1001/jama.2019.12085] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
IMPORTANCE The optimal international normalized ratio (INR) to prevent venous thromboembolism (VTE) in warfarin-treated patients with recent arthroplasty is unknown. OBJECTIVE To determine the safety and efficacy of a target INR of 1.8 vs 2.5 for VTE prophylaxis after orthopedic surgery. DESIGN, SETTING, AND PARTICIPANTS The randomized Genetic Informatics Trial (GIFT) of Warfarin to Prevent Deep Vein Thrombosis enrolled 1650 patients aged 65 years or older initiating warfarin for elective hip or knee arthroplasty at 6 US medical centers. Enrollment began in April 2011 and follow-up concluded in October 2016. INTERVENTIONS In a 2 × 2 factorial design, participants were randomized to a target INR of 1.8 (n = 823) or 2.5 (n = 827) and to either genotype-guided or clinically guided warfarin dosing. For the first 11 days of therapy, open-label warfarin dosing was guided by a web application. MAIN OUTCOMES AND MEASURES The primary outcome was the composite of VTE (within 60 days) or death (within 30 days). Participants underwent screening duplex ultrasound postoperatively. The hypothesis was that an INR target of 1.8 would be noninferior to an INR target of 2.5, using a noninferiority margin of 3% for the absolute risk of VTE. Secondary end points were bleeding and INR values of 4 or more. RESULTS Among 1650 patients who were randomized (mean age, 72.1 years; 1049 women [63.6%]; 1502 white [91.0%]), 1597 (96.8%) received at least 1 dose of warfarin and were included in the primary analysis. The rate of the primary composite outcome of VTE or death was 5.1% (41 of 804) in the low-intensity-warfarin group (INR target, 1.8) vs 3.8% (30 of 793) in the standard-treatment-warfarin group (INR target, 2.5), for a difference of 1.3% (1-sided 95% CI, -∞ to 3.05%, P = .06 for noninferiority). Major bleeding occurred in 0.4% of patients in the low-intensity group and 0.9% of patients in the standard-intensity group, for a difference of -0.5% (95% CI, -1.6% to 0.4%). The INR values of 4 or more occurred in 4.5% of patients in the low-intensity group and 12.2% of the standard-intensity group, for a difference of -7.8% (95% CI, -10.5% to -5.1%). CONCLUSIONS AND RELEVANCE Among older patients undergoing hip or knee arthroplasty and receiving warfarin prophylaxis, an international normalized ratio goal of 1.8 compared with 2.5 did not meet the criterion for noninferiority for risk of the composite outcome of VTE or death. However, the trial may have been underpowered to meet this criterion and further research may be warranted. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT01006733.
Collapse
Affiliation(s)
- Brian F. Gage
- Department of Medicine, Washington University in St Louis, St Louis, Missouri
| | - Anne R. Bass
- Department of Medicine, Hospital for Special Surgery, New York, New York
| | - Hannah Lin
- Department of Medicine, Washington University in St Louis, St Louis, Missouri
- Department of Medical Education, University of Massachusetts, Worcester
| | - Scott C. Woller
- Department of Medicine, Intermountain Medical Center, Salt Lake City, Utah
- Department of Medicine, University of Utah, Salt Lake City
| | - Scott M. Stevens
- Department of Medicine, Intermountain Medical Center, Salt Lake City, Utah
- Department of Medicine, University of Utah, Salt Lake City
| | - Noor Al-Hammadi
- Department of Medicine, Washington University in St Louis, St Louis, Missouri
| | - Jeffrey L. Anderson
- Department of Medicine, University of Utah, Salt Lake City
- Department of Cardiology, Intermountain Medical Center, Salt Lake City, Utah
| | - Juan Li
- Department of Medicine, Washington University in St Louis, St Louis, Missouri
| | - Tomás Rodriguez
- Department of Medicine, Washington University in St Louis, St Louis, Missouri
| | - J. Philip Miller
- Department of Medicine, Washington University in St Louis, St Louis, Missouri
| | | | | | - Amir K. Jaffer
- Department of Medicine, New York Presbyterian Queens Hospital, New York
| | - Cristi R. King
- Department of Medicine, Washington University in St Louis, St Louis, Missouri
| | - Brandi Whipple
- Department of Medicine, Washington University in St Louis, St Louis, Missouri
| | | | - Lynnae Napoli
- Department of Medicine, University of Utah, Salt Lake City
| | - Kerri Merritt
- Department of Medicine, Hospital for Special Surgery, New York, New York
| | - Anna M. Thompson
- Department of Medicine, Washington University in St Louis, St Louis, Missouri
- Department of Medical Education, University of Central Florida College of Medicine, Orlando
| | - Gina Hyun
- Department of Medicine, Washington University in St Louis, St Louis, Missouri
- Department of Radiology, Walter Reed National Military Medical Center, Bethesda, Maryland
| | - Wesley Hollomon
- Department of Orthopedic Surgery, Hospital for Special Surgery, New York, New York
| | - Robert L. Barrack
- Department of Orthopedic Surgery, Washington University in St Louis, St Louis, Missouri
| | - Ryan M. Nunley
- Department of Orthopedic Surgery, Washington University in St Louis, St Louis, Missouri
| | - Gerard Moskowitz
- Department of Medicine, Washington University in St Louis, St Louis, Missouri
| | - Victor Dávila-Román
- Department of Medicine, Washington University in St Louis, St Louis, Missouri
| | - Charles S. Eby
- Department of Medicine, Washington University in St Louis, St Louis, Missouri
- Department of Pathology and Immunology, Washington University in St Louis, St Louis, Missouri
| |
Collapse
|
111
|
Pratt VM, Cavallari LH, Del Tredici AL, Hachad H, Ji Y, Moyer AM, Scott SA, Whirl-Carrillo M, Weck KE. Recommendations for Clinical CYP2C9 Genotyping Allele Selection: A Joint Recommendation of the Association for Molecular Pathology and College of American Pathologists. J Mol Diagn 2019; 21:746-755. [PMID: 31075510 PMCID: PMC7057225 DOI: 10.1016/j.jmoldx.2019.04.003] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 04/03/2019] [Accepted: 04/22/2019] [Indexed: 01/17/2023] Open
Abstract
The goals of the Association for Molecular Pathology Pharmacogenomics (PGx) Working Group of the Association for Molecular Pathology Clinical Practice Committee are to define the key attributes of PGx alleles recommended for clinical testing and a minimum set of variants that should be included in clinical PGx genotyping assays. This document provides recommendations for a minimum panel of variant alleles (Tier 1) and an extended panel of variant alleles (Tier 2) that will aid clinical laboratories when designing assays for CYP2C9 testing. The Working Group considered the functional impact of the variants, allele frequencies in different populations and ethnicities, the availability of reference materials, and other technical considerations for PGx testing when developing these recommendations. Our goal is to promote standardization of testing PGx genes and alleles across clinical laboratories. These recommendations are not to be interpreted as restrictive but to provide a reference guide. The current document will focus on CYP2C9 testing that can be applied to all CYP2C9-related medications. A separate recommendation on warfarin PGx testing is being developed to include recommendations on CYP2C9 alleles and additional warfarin sensitivity-associated genes and alleles.
Collapse
Affiliation(s)
- Victoria M Pratt
- The Pharmacogenomics (PGx) Working Group of the Clinical Practice Committee, Association for Molecular Pathology, Bethesda, Maryland; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana.
| | - Larisa H Cavallari
- The Pharmacogenomics (PGx) Working Group of the Clinical Practice Committee, Association for Molecular Pathology, Bethesda, Maryland; Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida, Gainesville, Florida
| | - Andria L Del Tredici
- The Pharmacogenomics (PGx) Working Group of the Clinical Practice Committee, Association for Molecular Pathology, Bethesda, Maryland; Millennium Health, LLC, San Diego, California
| | - Houda Hachad
- The Pharmacogenomics (PGx) Working Group of the Clinical Practice Committee, Association for Molecular Pathology, Bethesda, Maryland; Translational Software, Bellevue, Washington
| | - Yuan Ji
- The Pharmacogenomics (PGx) Working Group of the Clinical Practice Committee, Association for Molecular Pathology, Bethesda, Maryland; Department of Pathology and ARUP Laboratories, University of Utah School of Medicine, Salt Lake City, Utah
| | - Ann M Moyer
- The Pharmacogenomics (PGx) Working Group of the Clinical Practice Committee, Association for Molecular Pathology, Bethesda, Maryland; Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Stuart A Scott
- The Pharmacogenomics (PGx) Working Group of the Clinical Practice Committee, Association for Molecular Pathology, Bethesda, Maryland; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York; Sema4, Stamford, Connecticut
| | - Michelle Whirl-Carrillo
- The Pharmacogenomics (PGx) Working Group of the Clinical Practice Committee, Association for Molecular Pathology, Bethesda, Maryland; Department of Biomedical Data Science, Stanford University, Stanford, California
| | - Karen E Weck
- The Pharmacogenomics (PGx) Working Group of the Clinical Practice Committee, Association for Molecular Pathology, Bethesda, Maryland; Department of Pathology and Laboratory Medicine and Department of Genetics, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
112
|
Bush WS, Cooke Bailey JN, Beno MF, Crawford DC. Bridging the Gaps in Personalized Medicine Value Assessment: A Review of the Need for Outcome Metrics across Stakeholders and Scientific Disciplines. Public Health Genomics 2019; 22:16-24. [PMID: 31454805 PMCID: PMC6752968 DOI: 10.1159/000501974] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 07/07/2019] [Indexed: 12/14/2022] Open
Abstract
Despite monumental advances in genomics, relatively few health care provider organizations in the United States offer personalized or precision medicine as part of the routine clinical workflow. The gaps between research and applied genomic medicine may be a result of a cultural gap across various stakeholders representing scientists, clinicians, patients, policy makers, and third party payers. Scientists are trained to assess the health care value of genomics by either quantifying population-scale effects, or through the narrow lens of clinical trials where the standard of care is compared with the predictive power of a single or handful of genetic variants. While these metrics are an essential first step in assessing and documenting the clinical utility of genomics, they are rarely followed up with other assessments of health care value that are critical to stakeholders who use different measures to define value. The limited value assessment in both the research and implementation science of precision medicine is likely due to necessary logistical constraints of these teams; engaging bioethicists, health care economists, and individual patient belief systems is incredibly daunting for geneticists and informaticians conducting research. In this narrative review, we concisely describe several definitions of value through various stakeholder viewpoints. We highlight the existing gaps that prevent clinical translation of scientific findings generally as well as more specifically using two present-day, extreme scenarios: (1) genetically guided warfarin dosing representing a handful of genetic markers and more than 10 years of basic and translational research, and (2) next-generation sequencing representing genome-dense data lacking substantial evidence for implementation. These contemporary scenarios highlight the need for various stakeholders to broadly adopt frameworks designed to define and collect multiple value measures across different disciplines to ultimately impact more universal acceptance of and reimbursement for genomic medicine.
Collapse
Affiliation(s)
- William S Bush
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Jessica N Cooke Bailey
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Mark F Beno
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Dana C Crawford
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio, USA,
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, USA,
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, USA,
| |
Collapse
|
113
|
Krebs K, Milani L. Translating pharmacogenomics into clinical decisions: do not let the perfect be the enemy of the good. Hum Genomics 2019; 13:39. [PMID: 31455423 PMCID: PMC6712791 DOI: 10.1186/s40246-019-0229-z] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 07/31/2019] [Indexed: 12/14/2022] Open
Abstract
The field of pharmacogenomics (PGx) is gradually shifting from the reactive testing of single genes toward the proactive testing of multiple genes to improve treatment outcomes, reduce adverse events, and decrease the burden of unnecessary costs for healthcare systems. Despite the progress in the field of pharmacogenomics, its implementation into routine care has been slow due to several barriers. However, in recent years, the number of studies on the implementation of PGx has increased, all providing a wealth of knowledge on different solutions for overcoming the obstacles that have been emphasized over the past years. This review focuses on some of the challenges faced by these initiatives, the solutions and different approaches for testing that they suggest, and the evidence that they provide regarding the benefits of preemptive PGx testing.
Collapse
Affiliation(s)
- Kristi Krebs
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Lili Milani
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| |
Collapse
|
114
|
Roden DM, McLeod HL, Relling MV, Williams MS, Mensah GA, Peterson JF, Van Driest SL. Pharmacogenomics. Lancet 2019; 394:521-532. [PMID: 31395440 PMCID: PMC6707519 DOI: 10.1016/s0140-6736(19)31276-0] [Citation(s) in RCA: 242] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 04/04/2019] [Accepted: 05/16/2019] [Indexed: 02/08/2023]
Abstract
Genomic medicine, which uses DNA variation to individualise and improve human health, is the subject of this Series of papers. The idea that genetic variation can be used to individualise drug therapy-the topic addressed here-is often viewed as within reach for genomic medicine. We have reviewed general mechanisms underlying variability in drug action, the role of genetic variation in mediating beneficial and adverse effects through variable drug concentrations (pharmacokinetics) and drug actions (pharmacodynamics), available data from clinical trials, and ongoing efforts to implement pharmacogenetics in clinical practice.
Collapse
Affiliation(s)
- Dan M Roden
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Howard L McLeod
- DeBartolo Family Personalized Medicine Institute, Moffitt Cancer Center, Tampa, FL, USA
| | - Mary V Relling
- Pharmaceutical Department, St Jude Children's Research Hospital, Memphis, TN, USA
| | | | - George A Mensah
- Center for Translation Research and Implementation Science, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Josh F Peterson
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sara L Van Driest
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
115
|
Musunuru K, Arora P, Cooke JP, Ferguson JF, Hershberger RE, Hickey KT, Lee JM, Lima JAC, Loscalzo J, Pereira NL, Russell MW, Shah SH, Sheikh F, Wang TJ, MacRae CA. Interdisciplinary Models for Research and Clinical Endeavors in Genomic Medicine: A Scientific Statement From the American Heart Association. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2019; 11:e000046. [PMID: 29844141 DOI: 10.1161/hcg.0000000000000046] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The completion of the Human Genome Project has unleashed a wealth of human genomics information, but it remains unclear how best to implement this information for the benefit of patients. The standard approach of biomedical research, with researchers pursuing advances in knowledge in the laboratory and, separately, clinicians translating research findings into the clinic as much as decades later, will need to give way to new interdisciplinary models for research in genomic medicine. These models should include scientists and clinicians actively working as teams to study patients and populations recruited in clinical settings and communities to make genomics discoveries-through the combined efforts of data scientists, clinical researchers, epidemiologists, and basic scientists-and to rapidly apply these discoveries in the clinic for the prediction, prevention, diagnosis, prognosis, and treatment of cardiovascular diseases and stroke. The highly publicized US Precision Medicine Initiative, also known as All of Us, is a large-scale program funded by the US National Institutes of Health that will energize these efforts, but several ongoing studies such as the UK Biobank Initiative; the Million Veteran Program; the Electronic Medical Records and Genomics Network; the Kaiser Permanente Research Program on Genes, Environment and Health; and the DiscovEHR collaboration are already providing exemplary models of this kind of interdisciplinary work. In this statement, we outline the opportunities and challenges in broadly implementing new interdisciplinary models in academic medical centers and community settings and bringing the promise of genomics to fruition.
Collapse
|
116
|
Helin TA, Joutsi-Korhonen L, Asmundela H, Niemi M, Orpana A, Lassila R. Warfarin dose requirement in patients having severe thrombosis or thrombophilia. Br J Clin Pharmacol 2019; 85:1684-1691. [PMID: 30933373 DOI: 10.1111/bcp.13948] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 03/12/2019] [Accepted: 03/23/2019] [Indexed: 12/17/2022] Open
Abstract
AIMS Warfarin dose requirement varies significantly. We compared the clinically established doses based on international normalized ratio (INR) among patients with severe thrombosis and/or thrombophilia with estimates from genetic dosing algorithms. METHODS Fifty patients with severe thrombosis and/or thrombophilia requiring permanent anticoagulation, referred to the Helsinki University Hospital Coagulation Center, were screened for thrombophilias and genotyped for CYP2C9*2 (c.430C>T, rs1799853), CYP2C9*3 (c.1075A>C, rs1057910) and VKORC1 c.-1639G>A (rs9923231) variants. The warfarin maintenance doses (target INR 2.0-3.0 in 94%, 2.5-3.5 in 6%) were estimated by the Gage and the International Warfarin Pharmacogenetics Consortium (IWPC) algorithms. The individual warfarin maintenance dose was tailored, supplementing estimates with comprehensive clinical evaluation and INR data. RESULTS Mean patient age was 47 years (range 20-76), and BMI 27 (SD 6), 68% being women. Forty-six (92%) had previous venous or arterial thrombosis, and 26 (52%) had a thrombophilia, with 22% having concurrent aspirin. A total of 40% carried the CYP2C9*2 or *3 allele and 54% carried the VKORC1-1639A allele. The daily mean maintenance dose of warfarin estimated by the Gage algorithm was 5.4 mg (95% CI 4.9-5.9 mg), and by the IWPC algorithm was 5.2 mg (95% CI 4.7-5.7 mg). The daily warfarin maintenance dose after clinical visits and follow-up was higher than the estimates, mean 6.9 mg (95% CI 5.6-8.2 mg, P < 0.006), with highest dose in patients having multiple thrombophilic factors (P < 0.03). CONCLUSIONS In severe thrombosis and/or thrombophilia, variation in thrombin generation and pharmacodynamics influences warfarin response. Pharmacogenetic dosing algorithms seem to underestimate dose requirement.
Collapse
Affiliation(s)
- Tuukka A Helin
- Coagulation Disorders Unit, Clinical Chemistry, University of Helsinki and HUSLAB, Helsinki University Hospital, Helsinki, Finland
| | - Lotta Joutsi-Korhonen
- Coagulation Disorders Unit, Clinical Chemistry, University of Helsinki and HUSLAB, Helsinki University Hospital, Helsinki, Finland
| | - Heidi Asmundela
- Coagulation Disorders Unit, Hematology, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
| | - Mikko Niemi
- Department of Clinical Pharmacology, Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki and HUSLAB, Helsinki University Hospital, Helsinki, Finland
| | - Arto Orpana
- Genetics and Clinical Chemistry, University of Helsinki and HUSLAB, Helsinki University Hospital, Helsinki, Finland
| | - Riitta Lassila
- Coagulation Disorders Unit, Clinical Chemistry, University of Helsinki and HUSLAB, Helsinki University Hospital, Helsinki, Finland.,Coagulation Disorders Unit, Hematology, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
117
|
Chanfreau-Coffinier C, Hull LE, Lynch JA, DuVall SL, Damrauer SM, Cunningham FE, Voight BF, Matheny ME, Oslin DW, Icardi MS, Tuteja S. Projected Prevalence of Actionable Pharmacogenetic Variants and Level A Drugs Prescribed Among US Veterans Health Administration Pharmacy Users. JAMA Netw Open 2019; 2:e195345. [PMID: 31173123 PMCID: PMC6563578 DOI: 10.1001/jamanetworkopen.2019.5345] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
IMPORTANCE Implementation of pharmacogenetic testing to guide drug prescribing has potential to improve drug response and prevent adverse events. Robust data exist for more than 30 gene-drug pairs linking genotype to drug response phenotypes; however, it is unclear which pharmacogenetic tests, if implemented, would provide the greatest utility for a given patient population. OBJECTIVES To project the proportion of veterans in the US Veterans Health Administration (VHA) with actionable pharmacogenetic variants and evaluate how testing might be associated with prescribing decisions. DESIGN, SETTING, AND PARTICIPANTS This cross-sectional study included veterans who used national VHA pharmacy services from October 1, 2011, to September 30, 2017. Data analyses began April 26, 2018, and were completed February 6, 2019. EXPOSURES Receipt of level A drugs based on VHA pharmacy dispensing records. MAIN OUTCOMES AND MEASURES Projected prevalence of actionable pharmacogenetic variants among VHA pharmacy users based on variant frequencies from the 1000 Genomes Project and veteran demographic characteristics; incident number of level A prescriptions, and proportion of new level A drug recipients projected to carry an actionable pharmacogenetic variant. RESULTS During the study, 7 769 359 veterans (mean [SD] age, 58.1 [17.8] years; 7 021 504 [90.4%] men) used VHA pharmacy services. It was projected that 99% of VHA pharmacy users would carry at least 1 actionable pharmacogenetic variant. Among VHA pharmacy users, 4 259 153 (54.8%) received at least 1 level A drug with 1 188 124 (15.3%) receiving 2 drugs, and 912 189 (11.7%) receiving 3 or more drugs. The most common incident prescriptions during the study were tramadol (923 671 new recipients), simvastatin (533 928 new recipients), citalopram (266 952 new recipients), and warfarin (205 177 new recipients). Gene-drug interactions projected to have substantial clinical impacts in the VHA population include the interaction of SLCO1B1 with simvastatin (1 988 956 veterans [25.6%]), CYP2D6 with tramadol (318 544 veterans [4.1%]), and CYP2C9 or VKORC1 with warfarin (7 163 349 veterans [92.2%]). CONCLUSIONS AND RELEVANCE Clinically important pharmacogenetic variants are highly prevalent in the VHA population. Almost all veterans would carry an actionable variant, and more than half of the population had been exposed to a drug affected by these variants. These results suggest that pharmacogenetic testing has the potential to affect pharmacotherapy decisions for commonly prescribed outpatient medications for many veterans.
Collapse
Affiliation(s)
- Catherine Chanfreau-Coffinier
- US Department of Veterans Affairs, VA Informatics and Computing Infrastructure, Salt Lake City Health Care System, Salt Lake City, Utah
| | - Leland E. Hull
- Center for Healthcare Organization and Implementation Research, US Department of Veterans Affairs, Boston Healthcare System, Boston, Massachusetts
- US Department of Veterans Affairs, Edith Nourse Rogers Memorial Veterans Hospital, Bedford, Massachusetts
| | - Julie A. Lynch
- US Department of Veterans Affairs, VA Informatics and Computing Infrastructure, Salt Lake City Health Care System, Salt Lake City, Utah
- US Department of Veterans Affairs, Edith Nourse Rogers Memorial Veterans Hospital, Bedford, Massachusetts
- College of Nursing and Health Sciences, University of Massachusetts, Boston
| | - Scott L. DuVall
- US Department of Veterans Affairs, VA Informatics and Computing Infrastructure, Salt Lake City Health Care System, Salt Lake City, Utah
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City
| | - Scott M. Damrauer
- Perelman School of Medicine, University of Pennsylvania, Philadelphia
- Corporal Michael Crescenz Department of Veterans Affairs Medical Center, Philadelphia, Pennsylvania
| | - Francesca E. Cunningham
- US Department of Veterans Affairs Center for Medication Safety, Pharmacy Benefits Management Services, Hines, Illinois
| | | | - Michael E. Matheny
- Geriatrics Research Education and Clinical Care Center, US Department of Veterans Affairs Tennessee Valley Healthcare System, Nashville
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - David W. Oslin
- Perelman School of Medicine, University of Pennsylvania, Philadelphia
- Corporal Michael Crescenz Department of Veterans Affairs Medical Center, Philadelphia, Pennsylvania
| | - Michael S. Icardi
- US Department of Veterans Affairs Iowa City Healthcare System, Iowa City, Iowa
- US Department of Veterans Affairs National Office of Pathology and Laboratory Medicine, Iowa City, Iowa
- Carver College of Medicine, University of Iowa, Iowa City
| | - Sony Tuteja
- Perelman School of Medicine, University of Pennsylvania, Philadelphia
- Corporal Michael Crescenz Department of Veterans Affairs Medical Center, Philadelphia, Pennsylvania
| |
Collapse
|
118
|
Lin B, Chung WK. Cases in Precision Medicine: The Role of Pharmacogenetics in Precision Prescribing. Ann Intern Med 2019; 170:796-804. [PMID: 31108507 PMCID: PMC7458588 DOI: 10.7326/m18-2357] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Pharmacogenetics may help physicians deliver individualized treatments based on how a person's genes affect a drug's effects and metabolism. This information can help prevent adverse events or improve drug efficacy by enabling the physician to optimize dosage or to avoid a medication with adverse reactions and to prescribe an alternative therapy. This article discusses the current clinical utility of pharmacogenetic testing in the context of a patient who requires anticoagulation with warfarin.
Collapse
Affiliation(s)
- Bohan Lin
- Columbia University, New York, New York (B.L., W.K.C.)
| | - Wendy K Chung
- Columbia University, New York, New York (B.L., W.K.C.)
| |
Collapse
|
119
|
Fitzgerald G, Prince C, Downing J, Reynolds J, Zhang JE, Hanson A, Alfirevic A, Pirmohamed M. Processes and barriers to implementation of point-of-care genotype-guided dosing of warfarin into UK outpatient anticoagulation clinics. Pharmacogenomics 2019; 20:599-608. [DOI: 10.2217/pgs-2019-0021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Gail Fitzgerald
- Research Nurse, Royal Liverpool & Broadgreen University Hospitals NHS Trust, Wolfson Centre for Personalised Medicine, University of Liverpool, Liverpool, UK
| | - Clare Prince
- Research Nurse, Royal Liverpool & Broadgreen University Hospitals NHS Trust, Wolfson Centre for Personalised Medicine, University of Liverpool, Liverpool, UK
| | - Jennifer Downing
- Research Manager, NIHR CLAHRC NWC, University of Liverpool, Liverpool, UK
| | - Julia Reynolds
- Associate Director, Innovation Agency, Academic Health Science Network for the North West Coast, Warrington, UK
| | - Jieying Eunice Zhang
- Post-doctoral Research Associate, Wolfson Centre for Personalised Medicine, University of Liverpool, Liverpool, UK
| | - Anita Hanson
- Research Nurse, Royal Liverpool & Broadgreen University Hospitals NHS Trust, Wolfson Centre for Personalised Medicine, University of Liverpool, Liverpool, UK
| | - Ana Alfirevic
- Professor of Pharmacology, University of Liverpool, Liverpool, UK
| | - Munir Pirmohamed
- David Weatherall Chair of Medicine, University of Liverpool & Royal Liverpool & Broadgreen University Hospitals NHS Trust, Liverpool, UK
| |
Collapse
|
120
|
Danese E, Raimondi S, Montagnana M, Tagetti A, Langaee T, Borgiani P, Ciccacci C, Carcas AJ, Borobia AM, Tong HY, Dávila-Fajardo C, Botton MR, Bourgeois S, Deloukas P, Caldwell MD, Burmester JK, Berg RL, Cavallari LH, Drozda K, Huang M, Zhao LZ, Cen HJ, Gonzalez-Conejero R, Roldan V, Nakamura Y, Mushiroda T, Gong IY, Kim RB, Hirai K, Itoh K, Isaza C, Beltrán L, Jiménez-Varo E, Cañadas-Garre M, Giontella A, Kringen MK, Foss Haug KB, Gwak HS, Lee KE, Minuz P, Lee MTM, Lubitz SA, Scott S, Mazzaccara C, Sacchetti L, Genç E, Özer M, Pathare A, Krishnamoorthy R, Paldi A, Siguret V, Loriot MA, Kutala VK, Suarez-Kurtz G, Perini J, Denny JC, Ramirez AH, Mittal B, Rathore SS, Sagreiya H, Altman R, Shahin MHA, Khalifa SI, Limdi NA, Rivers C, Shendre A, Dillon C, Suriapranata IM, Zhou HH, Tan SL, Tatarunas V, Lesauskaite V, Zhang Y, Maitland-van der Zee AH, Verhoef TI, de Boer A, Taljaard M, Zambon CF, Pengo V, Zhang JE, Pirmohamed M, Johnson JA, Fava C. Effect of CYP4F2, VKORC1, and CYP2C9 in Influencing Coumarin Dose: A Single-Patient Data Meta-Analysis in More Than 15,000 Individuals. Clin Pharmacol Ther 2019; 105:1477-1491. [PMID: 30506689 PMCID: PMC6542461 DOI: 10.1002/cpt.1323] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 11/18/2018] [Indexed: 11/06/2022]
Abstract
The cytochrome P450 (CYP)4F2 gene is known to influence mean coumarin dose. The aim of the present study was to undertake a meta-analysis at the individual patients level to capture the possible effect of ethnicity, gene-gene interaction, or other drugs on the association and to verify if inclusion of CYP4F2*3 variant into dosing algorithms improves the prediction of mean coumarin dose. We asked the authors of our previous meta-analysis (30 articles) and of 38 new articles retrieved by a systematic review to send us individual patients' data. The final collection consists of 15,754 patients split into a derivation and validation cohort. The CYP4F2*3 polymorphism was consistently associated with an increase in mean coumarin dose (+9% (95% confidence interval (CI) 7-10%), with a higher effect in women, in patients taking acenocoumarol, and in white patients. The inclusion of the CYP4F2*3 in dosing algorithms slightly improved the prediction of stable coumarin dose. New pharmacogenetic equations potentially useful for clinical practice were derived.
Collapse
Affiliation(s)
- Elisa Danese
- Clinical Biochemistry Section, Department of Neurological, Biomedical and Movement Sciences, University of Verona, Verona, Italy
| | - Sara Raimondi
- General Medicine and Hypertension Unit, Department of Medicine, University of Verona, Verona, Italy
| | - Martina Montagnana
- Clinical Biochemistry Section, Department of Neurological, Biomedical and Movement Sciences, University of Verona, Verona, Italy
| | - Angela Tagetti
- General Medicine and Hypertension Unit, Department of Medicine, University of Verona, Verona, Italy
| | - Taimour Langaee
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Paola Borgiani
- Genetics Section, Department of Biomedicine and Prevention, University of Rome “Tor Vergata,” Rome, Italy
| | - Cinzia Ciccacci
- Genetics Section, Department of Biomedicine and Prevention, University of Rome “Tor Vergata,” Rome, Italy
| | - Antonio J. Carcas
- Clinical Pharmacology Department, La Paz University Hospital, School of Medicine, IdiPAZ, Universidad Autónoma de Madrid, Madrid, Spain
- Spanish Clinical Research Network-SCReN, Madrid, Spain
| | - Alberto M. Borobia
- Clinical Pharmacology Department, La Paz University Hospital, School of Medicine, IdiPAZ, Universidad Autónoma de Madrid, Madrid, Spain
- Spanish Clinical Research Network-SCReN, Madrid, Spain
| | - Hoi Y. Tong
- Clinical Pharmacology Department, La Paz University Hospital, School of Medicine, IdiPAZ, Universidad Autónoma de Madrid, Madrid, Spain
- Spanish Clinical Research Network-SCReN, Madrid, Spain
| | - Cristina Dávila-Fajardo
- Department of Clinical Pharmacy, San Cecilio University Hospital, Institute for Biomedical Research, IBS, Granada, Spain
| | | | - Stephane Bourgeois
- William Harvey Research Institute, Barts & the London Medical School, Queen Mary University of London, London, UK
| | - Panos Deloukas
- William Harvey Research Institute, Barts & the London Medical School, Queen Mary University of London, London, UK
- Princess Al-Jawhara Al-Brahim Centre of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Michael D. Caldwell
- Center for Hyperbaric Medicine and Tissue Repair, Marshfield Clinic, Marshfield, Wisconsin, USA
| | - Jim K. Burmester
- Grants Office, Gundersen Health System, La Crosse, Wisconsin, USA
| | - Richard L. Berg
- Clinical Research Center, Marshfield Clinic Research Foundation, Marshfield, Wisconsin, USA
| | - Larisa H. Cavallari
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Katarzyna Drozda
- Department of Pharmacy Practice, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Min Huang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Li-Zi Zhao
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Han-Jing Cen
- Guangzhou Women and Children’s Medical Center, Guangzhou, China
| | - Rocio Gonzalez-Conejero
- Centro Regional de Hemodonación, Hospital Universitario Morales Meseguer, Universidad de Murcia, Murcia, Spain
| | - Vanessa Roldan
- Centro Regional de Hemodonación, Hospital Universitario Morales Meseguer, Universidad de Murcia, Murcia, Spain
| | - Yusuke Nakamura
- Research Group for Pharmacogenomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Taisei Mushiroda
- Research Group for Pharmacogenomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Inna Y. Gong
- Division of Clinical Pharmacology, Department of Medicine, University of Western Ontario, London, Ontario, Canada
| | - Richard B. Kim
- Division of Clinical Pharmacology, Department of Medicine, University of Western Ontario, London, Ontario, Canada
| | - Keita Hirai
- Department of Clinical Pharmacology & Genetics, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Kunihiko Itoh
- Department of Clinical Pharmacology & Genetics, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Carlos Isaza
- Faculty of Heath Sciences, Laboratory of Medical Genetics, Universidad Tecnológica de Pereira, Pereira, Colombia
| | - Leonardo Beltrán
- Faculty of Heath Sciences, Laboratory of Medical Genetics, Universidad Tecnológica de Pereira, Pereira, Colombia
- Faculty of Heath Sciences, Unidad Central del Valle del Cauca, Valle del Cauca, Colombia
| | | | - Marisa Cañadas-Garre
- Centre for Public Health, School of Medicine, Dentistry, and Biomedical Sciences, Queen’s University Belfast, Belfast, UK
| | - Alice Giontella
- General Medicine and Hypertension Unit, Department of Medicine, University of Verona, Verona, Italy
| | - Marianne K. Kringen
- Department of Pharmacology, Oslo University Hospital, Ullevål, Oslo, Norway
- Center for Psychopharmacology, Diakonhjemmet Hospital, Oslo, Norway
| | - Kari Bente Foss Haug
- Department of Medical Biochemistry, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Hye Sun Gwak
- Division of Life and Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Kyung Eun Lee
- College of Pharmacy, Chungbuk National University, Cheongju-si, Korea
| | - Pietro Minuz
- General Medicine and Hypertension Unit, Department of Medicine, University of Verona, Verona, Italy
| | - Ming Ta Michael Lee
- Genomic Medicine Institute, Geisinger Health System, Danville, Pennsylvania, USA
- National Center for Genome Medicine, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Steven A. Lubitz
- Cardiac Arrhythmia Service & Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Stuart Scott
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Cristina Mazzaccara
- CEINGE–Biotecnologie Avanzate s.c.ar.l., Napoli, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Napoli, Italy
| | - Lucia Sacchetti
- CEINGE–Biotecnologie Avanzate s.c.ar.l., Napoli, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Napoli, Italy
| | - Ece Genç
- Department of Pharmacology, Yeditepe University, Istanbul, Turkey
| | - Mahmut Özer
- Department of Pharmacology, Yeditepe University, Istanbul, Turkey
| | - Anil Pathare
- College of Medicine & Health Sciences, Sultan Qaboos University, Muscat, Oman
| | | | - Andras Paldi
- Ecole Pratique des Hautes Etudes, UMRS_951, Genethon, Evry, France
| | - Virginie Siguret
- Sorbonne Paris Cité, INSERM, UMR-S-1140, Université Paris Descartes, Paris, France
- Assistance Publique Hôpitaux de Paris, Hôpital Lariboisière, Service d’Hématologie Biologique, Paris, France
| | - Marie-Anne Loriot
- Sorbonne Paris Cité, INSERM, UMR-S-1147, Université Paris Descartes, Paris, France
- Assistance Publique Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Biochimie UF Pharmacogénétique et Oncologie Moléculaire, Paris, France
| | - Vijay Kumar Kutala
- Department of Clinical Pharmacology & Therapeutics, Nizam’s Institute of Medical Sciences, Hyderabad, India
| | | | - Jamila Perini
- Research Laboratory of Pharmaceutical Sciences, West Zone State University-UEZO, Rio de Janeiro, Brazil
| | - Josh C. Denny
- Department of Medicine and Department of Biomedical Informatics, Vanderbilt University, Nashville, Tennessee, USA
| | - Andrea H. Ramirez
- Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Balraj Mittal
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | | | - Hersh Sagreiya
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA
| | - Russ Altman
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA
| | - Mohamed Hossam A. Shahin
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Sherief I. Khalifa
- College of Pharmacy, Gulf Medical University, Ajman, United Arab Emirates
| | - Nita A. Limdi
- Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Charles Rivers
- Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Aditi Shendre
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indiana University Purdue University, Indianapolis, Indiana, USA
| | - Chrisly Dillon
- Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Ivet M. Suriapranata
- Mochtar Riady Institute for Nanotechnology, Universitas Pelita Harapan, Lippo Karawaci, Tangerang, Banten, Indonesia
| | - Hong-Hao Zhou
- Institute of Clinical Pharmacology, Central South University, Hunan Sheng, China
| | - Sheng-Lan Tan
- Department of Pharmacy, Xiangya Second Hospital, Central South University, Hunan Sheng, China
| | - Vacis Tatarunas
- Laboratory of Molecular Cardiology, Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Vaiva Lesauskaite
- Laboratory of Molecular Cardiology, Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Yumao Zhang
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Anke H. Maitland-van der Zee
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
- Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Talitha I. Verhoef
- Department of Applied Health Research, University College London, London, UK
| | - Anthonius de Boer
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Monica Taljaard
- Clinica Epidemiology Program and Department of Epidemiology and Community Medicine, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | | | - Vittorio Pengo
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Padua, Italy
| | - Jieying Eunice Zhang
- Wolfson Centre for Personalised Medicine, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Munir Pirmohamed
- Wolfson Centre for Personalised Medicine, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Julie A. Johnson
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Cristiano Fava
- General Medicine and Hypertension Unit, Department of Medicine, University of Verona, Verona, Italy
| |
Collapse
|
121
|
Kampouraki E, Kamali F. Pharmacogenetics of anticoagulants used for stroke prevention in patients with atrial fibrillation. Expert Opin Drug Metab Toxicol 2019; 15:449-458. [PMID: 31120800 DOI: 10.1080/17425255.2019.1623878] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Introduction: The inclusion of pharmacogenetics alongside clinical information in anticoagulant therapy offers the opportunity for a tailored approach to treatment according to individual patient characteristics. Areas covered: Literature was searched using PubMed database, focusing on pharmacogenetics of oral anticoagulants. Original research articles and review articles in English language were included in the literature reviewed. This article includes all information available for the genetic cause of inter-individual variability in anticoagulation response to oral anticoagulant drugs. The pharmacogenetics of VKAs and NOACs are described in detail. Expert opinion: There have been numerous studies focusing on the pharmacogenetics of VKAs, particularly warfarin. Current evidence suggests that known genetic and clinical factors explain a large proportion of the inter-individual variability in response to warfarin. Pharmacogenetic-based algorithms have been validated to determine their clinical utility with equivocal results. To date, only a limited number of mostly small studies on the pharmacogenetics of NOACs exists. The latter have highlighted genetic polymorphisms in specific genes that may affect clinical outcomes. Further evaluations of these polymorphisms are needed before firm conclusions can be drawn about the significance of pharmacogenetics on NOAC therapy.
Collapse
Affiliation(s)
- Emmanouela Kampouraki
- a Institute of Cellular Medicine within Faculty of Medical Sciences , Newcastle University , Newcastle upon Tyne , UK
| | - Farhad Kamali
- b Newcastle upon Tyne Hospitals, NHS Foundation Trust , Newcastle upon Tyne , UK
| |
Collapse
|
122
|
Benincasa G, Costa D, Infante T, Lucchese R, Donatelli F, Napoli C. Interplay between genetics and epigenetics in modulating the risk of venous thromboembolism: A new challenge for personalized therapy. Thromb Res 2019; 177:145-153. [DOI: 10.1016/j.thromres.2019.03.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 02/12/2019] [Accepted: 03/13/2019] [Indexed: 02/09/2023]
|
123
|
Cao X, Chen S, Wang Z, Liu Y, Luan X, Hou S, Li W, Shi H. The label-free detection and distinction of CYP2C9-expressing and non-expressing cells by surface-enhanced Raman scattering substrates based on bimetallic AuNPs-AgNWs. RSC Adv 2019; 9:13304-13315. [PMID: 35520768 PMCID: PMC9063916 DOI: 10.1039/c9ra02046b] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 04/18/2019] [Indexed: 11/21/2022] Open
Abstract
Cytochrome P450 2C9 (CYP2C9) is capable of catalyzing the biotransformation of endogenous compounds in cells, indicating that this enzyme could change the intracellular environment and is related to the pathogenesis of diseases. Currently, it is still a challenge to study the differences in cellular components between CYP2C9-expressing and non-expressing cells. In this study, employing a Au nanoparticles-Ag nanowires (AuNPs-AgNWs) decorated silicon wafer as a novel non-destructive and label-free tool, we applied surface-enhanced Raman scattering (SERS) spectroscopy to detect and distinguish the cellular composition of CYP2C9-expressing cells (293T-Mig-2C9) and non-expressing cells (293T-Mig-R1). AgNWs with high surface roughness were formed by modification of AuNPs onto their surface by electrostatic interactions, which enabled them to exhibit greatly enhanced SERS ability. Then, they were employed to fabricate SERS substrates via an electrostatically assisted 3-aminopropyltriethoxysilane (APTES)-functionalized surface-assembly method. The SERS substrates exhibited high sensitivity with a detection limit of 1 × 10-9 M for 4-mercaptobenzoic acid (4-MBA). Meanwhile, the SERS substrates exhibited good uniformity and reproducibility. The cytotoxicity assay demonstrated that the SERS substrates displayed good biocompatibility with 293T cells. Before SERS measurements, CYP2C9 constantly expressed cells (293T-Mig-2C9 cells) and control cells (293T-Mig-R1 cells) were constructed. The expression of CYP2C9 and the catalytic activity in the cells were checked. Using the AuNPs-AgNWs substrates as a high-performance in vitro sensing platform allowed us to obtain fingerprint spectra of 293T-Mig-R1 and 293T-Mig-2C9 cells. The difference spectra between the two cell lines were studied to interpret the spectral differences and gain insight into the biochemical variations. Finally, principal component analysis (PCA) score plots of the SERS spectra were also used to better view the differences between the two cell lines. SERS detection based on the AuNPs-AgNWs substrates provides a sensitive, non-destructive and label-free method for differentiation between 293T-Mig-R1 and 293T-Mig-2C9 cells.
Collapse
Affiliation(s)
- Xiaowei Cao
- Institute of Translational Medicine, Medical College, Yangzhou University Yangzhou 225001 PR China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University Yangzhou 225001 PR China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University Yangzhou 225009 China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine Yangzhou 225001 PR China
| | - Shuai Chen
- Institute of Translational Medicine, Medical College, Yangzhou University Yangzhou 225001 PR China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University Yangzhou 225009 China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research Yangzhou 225001 PR China
| | - Zhenyu Wang
- Institute of Translational Medicine, Medical College, Yangzhou University Yangzhou 225001 PR China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University Yangzhou 225009 China
| | - Yong Liu
- School of Life Science and Medicine, Dalian University of Technology Panjin 124221 China
| | - Xiaowei Luan
- School of Life Science and Medicine, Dalian University of Technology Panjin 124221 China
| | - Sicong Hou
- Institute of Translational Medicine, Medical College, Yangzhou University Yangzhou 225001 PR China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University Yangzhou 225001 PR China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University Yangzhou 225009 China
| | - Wei Li
- Institute of Translational Medicine, Medical College, Yangzhou University Yangzhou 225001 PR China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research Yangzhou 225001 PR China
| | - Hongcan Shi
- Institute of Translational Medicine, Medical College, Yangzhou University Yangzhou 225001 PR China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University Yangzhou 225001 PR China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research Yangzhou 225001 PR China
| |
Collapse
|
124
|
Caudle KE, Gammal RS, Karnes JH, Afanasjeva J, Anderson KC, Barreto EF, Beavers C, Bhat S, Birrer KL, Chahine EB, Ensor CR, Flowers SA, Formea CM, George JM, Gosser RA, Hebert MF, Karaoui LR, Kolpek JH, Lee JC, Leung JG, Maldonado AQ, Minze MG, Pulk RA, Shelton CM, Sheridan M, Smith MA, Soefje S, Tellez-Corrales E, Walko CM, Cavallari LH. PRN OPINION PAPER: Application of precision medicine across pharmacy specialty areas. JOURNAL OF THE AMERICAN COLLEGE OF CLINICAL PHARMACY 2019. [DOI: 10.1002/jac5.1107] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Kelly E. Caudle
- Department of Pharmaceutical Sciences; St. Jude Children's Research Hospital; Memphis Tennessee
| | - Roseann S. Gammal
- Department of Pharmaceutical Sciences; St. Jude Children's Research Hospital; Memphis Tennessee
- Department of Pharmacy Practice; MCPHS University School of Pharmacy; Boston Massachusetts
| | - Jason H. Karnes
- Department of Pharmacy Practice and Science; University of Arizona College of Pharmacy; Tucson Arizona
| | - Janna Afanasjeva
- Drug Information Group; University of Illinois College of Pharmacy; Chicago Illinois
| | | | - Erin F. Barreto
- Department of Pharmacy; Mayo Clinic; Rochester Minnesota
- Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery; Mayo Clinic; Rochester Minnesota
| | - Craig Beavers
- Department of Pharmacy Service; University of Kentucky Healthcare; Lexington Kentucky
- Department of Pharmacy Practice & Science; University of Kentucky College of Pharmacy; Lexington Kentucky
| | - Shubha Bhat
- Department of Pharmacy; Boston Medical Center; Boston Massachusetts
| | - Kara L. Birrer
- Pharmacy Services, Orlando Regional Medical Center/Orlando Health; Orlando Florida
| | - Elias B. Chahine
- Department of Pharmacy Practice; Palm Beach Atlantic University Lloyd L. Gregory School of Pharmacy; West Palm Beach Florida
| | | | - Stephanie A. Flowers
- Department of Pharmacy Practice; University of Illinois at Chicago; Chicago Illinois
| | | | - Jomy M. George
- Clinical Pharmacokinetics Research Unit, Clinical Center Pharmacy; National Institutes of Health; Bethesda Maryland
| | - Rena A. Gosser
- Department of Pharmacy; University of Washington Medicine; Seattle Washington
| | - Mary F. Hebert
- Departments of Pharmacy and Obstetrics & Gynecology; University of Washington; Seattle Washington
| | - Lamis R. Karaoui
- Department of Pharmacy Practice; Lebanese American University School of Pharmacy; Byblos Lebanon
| | - Jimmi Hatton Kolpek
- Department of Pharmacy Practice & Science; University of Kentucky College of Pharmacy; Lexington Kentucky
| | - James C. Lee
- Department of Pharmacy Practice; University of Illinois at Chicago; Chicago Illinois
| | | | - Angela Q. Maldonado
- Department of Transplant Surgery; Vidant Medical Center; Greenville North Carolina
| | - Molly G. Minze
- Department of Pharmacy Practice; Texas Tech University Health Sciences Center School of Pharmacy; Abilene Texas
| | - Rebecca A. Pulk
- Corporate Pharmacy Services; Yale New Haven Health; New Haven Connecticut
| | - Chasity M. Shelton
- Department of Clinical Pharmacy and Translational Science; The University of Tennessee Health Science Center; Memphis Tennessee
| | | | - Michael A. Smith
- Department of Clinical Pharmacy; University of Michigan; Ann Arbor Michigan
| | - Scott Soefje
- Department of Pharmacy Services; Mayo Clinic; Rochester Minnesota
| | - Eglis Tellez-Corrales
- Department Pharmacy Practice, College of Pharmacy; Marshall B Ketchum University; Fullerton California
| | - Christine M. Walko
- DeBartolo Family Personalized Medicine Institute, Moffitt Cancer Center; Tampa Florida
- Department of Oncologic Sciences, Morsani College of Medicine; University of South Florida; Tampa Florida
| | - Larisa H. Cavallari
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics; University of Florida; Gainesville Florida
| |
Collapse
|
125
|
Jorgensen AL, Prince C, Fitzgerald G, Hanson A, Downing J, Reynolds J, Zhang JE, Alfirevic A, Pirmohamed M. Implementation of genotype-guided dosing of warfarin with point-of-care genetic testing in three UK clinics: a matched cohort study. BMC Med 2019; 17:76. [PMID: 30961588 PMCID: PMC6454722 DOI: 10.1186/s12916-019-1308-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 03/15/2019] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Warfarin is a widely used oral anticoagulant. Determining the correct dose required to maintain the international normalised ratio (INR) within a therapeutic range can be challenging. In a previous trial, we showed that a dosing algorithm incorporating point-of-care genotyping information ('POCT-GGD' approach) led to improved anticoagulation control. To determine whether this approach could translate into clinical practice, we undertook an implementation project using a matched cohort design. METHODS At three clinics (implementation group; n = 119), initial doses were calculated using the POCT-GGD approach; at another three matched clinics (control group; n = 93), patients were dosed according to the clinic's routine practice. We also utilised data on 640 patients obtained from routinely collected data at comparable clinics. Primary outcome was percentage time in target INR range. Patients and staff from the implementation group also provided questionnaire feedback on POCT-GGD. RESULTS Mean percentage time in INR target range was 55.25% in the control group and 62.74% in the implementation group; therefore, 7.49% (95% CI 3.41-11.57%) higher in the implementation group (p = 0.0004). Overall, patients and staff viewed POCT-GGD positively, suggesting minor adjustments to allow smooth implementation into practice. CONCLUSIONS In the first demonstration of the implementation of genotype-guided dosing, we show that warfarin dosing determined using an algorithm incorporating genetic and clinical factors can be implemented smoothly into clinic, to ensure target INR range is reached sooner and maintained. The findings are like our previous randomised controlled trial, providing an alternative method for improving the risk-benefit of warfarin use in daily practice.
Collapse
Affiliation(s)
- Andrea L Jorgensen
- Department of Biostatistics, Institute of Translational Medicine, University of Liverpool, member of Liverpool Health Partners, Liverpool, UK.
| | - Clare Prince
- The Royal Liverpool and Broadgreen University Hospitals NHS Trust and Wolfson Centre for Personalised Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Gail Fitzgerald
- The Royal Liverpool and Broadgreen University Hospitals NHS Trust and Wolfson Centre for Personalised Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Anita Hanson
- The Royal Liverpool and Broadgreen University Hospitals NHS Trust and Wolfson Centre for Personalised Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Jennifer Downing
- Wolfson Centre for Personalised Medicine, Institute of Translational Medicine, University of Liverpool, member of Liverpool Health Partners, Liverpool, UK.,NIHR Collaboration for Leadership in Applied Health Research and Care, North West Coast, UK
| | - Julia Reynolds
- Innovation Agency, Academic Health Science Network for the North West Coast, Daresbury, Warrington, UK
| | - J Eunice Zhang
- Wolfson Centre for Personalised Medicine, Institute of Translational Medicine, University of Liverpool, member of Liverpool Health Partners, Liverpool, UK
| | - Ana Alfirevic
- Wolfson Centre for Personalised Medicine, Institute of Translational Medicine, University of Liverpool, member of Liverpool Health Partners, Liverpool, UK
| | - Munir Pirmohamed
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool and The Royal Liverpool and Broadgreen University Hospitals NHS Trust, members of Liverpool Health Partners, Liverpool, UK
| |
Collapse
|
126
|
Bonney PA, Yim B, Brinjikji W, Walcott BP. Pharmacogenomic considerations for antiplatelet agents: the era of precision medicine in stroke prevention and neurointerventional practice. Cold Spring Harb Mol Case Stud 2019; 5:mcs.a003731. [PMID: 30936195 PMCID: PMC6549574 DOI: 10.1101/mcs.a003731] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Antiplatelet drugs are widely utilized in the setting of primary stroke prevention, secondary stroke prevention, and neuroendovascular device-related stroke prevention. These medications are effective in general, although significant variability in drug activity exists between patients. Although this variation may be related in part to a multitude of factors, a growing body of evidence suggests that individual genotypes are a main contributor. The PharmGKB database was mined to prioritize genetic variants with potential clinical relevance for response to aspirin, clopidogrel, prasugrel, and ticagrelor. Although variants were reported for all drugs, the highest level of evidence was found in cytochrome P450 (CYP450) genotype variation related to clopidogrel response. Individual genetic influences have an impact on the pharmacodynamics of antiplatelet agents. Current clinical practice for stroke prevention is primarily empiric or guided by functional assays; however, there now exists a third potential pathway to base treatment decisions: genotype-guided treatment.
Collapse
Affiliation(s)
- Phillip A Bonney
- Department of Neurological Surgery, University of Southern California, Los Angeles, California 90033, USA
| | - Benjamin Yim
- Department of Neurological Surgery, University of Southern California, Los Angeles, California 90033, USA
| | - Waleed Brinjikji
- Departments of Neurosurgery and Radiology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Brian P Walcott
- Department of Neurological Surgery, University of Southern California, Los Angeles, California 90033, USA
| |
Collapse
|
127
|
Dávila-Fajardo CL, Díaz-Villamarín X, Antúnez-Rodríguez A, Fernández-Gómez AE, García-Navas P, Martínez-González LJ, Dávila-Fajardo JA, Barrera JC. Pharmacogenetics in the Treatment of Cardiovascular Diseases and Its Current Progress Regarding Implementation in the Clinical Routine. Genes (Basel) 2019; 10:genes10040261. [PMID: 30939847 PMCID: PMC6523655 DOI: 10.3390/genes10040261] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 03/25/2019] [Accepted: 03/26/2019] [Indexed: 12/18/2022] Open
Abstract
There is a special interest in the implementation of pharmacogenetics in clinical practice, although there are some barriers that are preventing this integration. A large part of these pharmacogenetic tests are focused on drugs used in oncology and psychiatry fields and for antiviral drugs. However, the scientific evidence is also high for other drugs used in other medical areas, for example, in cardiology. In this article, we discuss the evidence and guidelines currently available on pharmacogenetics for clopidogrel, warfarin, acenocoumarol, and simvastatin and its implementation in daily clinical practice.
Collapse
Affiliation(s)
- Cristina Lucía Dávila-Fajardo
- Department of Clinical Pharmacy, San Cecilio University Hospital, Institute for Biomedical Research, ibs.GRANADA, 18016 Granada, Spain.
| | - Xando Díaz-Villamarín
- Department of Clinical Pharmacy, San Cecilio University Hospital, Institute for Biomedical Research, ibs.GRANADA, 18016 Granada, Spain.
| | - Alba Antúnez-Rodríguez
- Genomics Unit, Centro Pfizer-Universidad de Granada-Junta de Andalucía de Genómica e Investigación Oncológica (Genyo), 18016 Granada, Spain.
| | - Ana Estefanía Fernández-Gómez
- Department of Clinical Pharmacy, San Cecilio University Hospital, Institute for Biomedical Research, ibs.GRANADA, 18016 Granada, Spain.
| | - Paloma García-Navas
- Department of Clinical Pharmacy, San Cecilio University Hospital, Institute for Biomedical Research, ibs.GRANADA, 18016 Granada, Spain.
| | - Luis Javier Martínez-González
- Genomics Unit, Centro Pfizer-Universidad de Granada-Junta de Andalucía de Genómica e Investigación Oncológica (Genyo), 18016 Granada, Spain.
| | | | - José Cabeza Barrera
- Department of Clinical Pharmacy, San Cecilio University Hospital, Institute for Biomedical Research, ibs.GRANADA, 18016 Granada, Spain.
| |
Collapse
|
128
|
Langaee T, El Rouby N, Stauffer L, Galloway C, Cavallari LH. Development and Cross-Validation of High-Resolution Melting Analysis-Based Cardiovascular Pharmacogenetics Genotyping Panel. Genet Test Mol Biomarkers 2019; 23:209-214. [DOI: 10.1089/gtmb.2018.0298] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Taimour Langaee
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, Florida
| | - Nihal El Rouby
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, Florida
| | - Lynda Stauffer
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, Florida
| | - Cheryl Galloway
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, Florida
| | - Larisa H. Cavallari
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, Florida
| |
Collapse
|
129
|
Differences in Warfarin Pharmacodynamics and Predictors of Response Among Three Racial Populations. Clin Pharmacokinet 2019; 58:1077-1089. [DOI: 10.1007/s40262-019-00745-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
130
|
Li X, Li D, Wu JC, Liu ZQ, Zhou HH, Yin JY. Precision dosing of warfarin: open questions and strategies. THE PHARMACOGENOMICS JOURNAL 2019; 19:219-229. [PMID: 30745565 DOI: 10.1038/s41397-019-0083-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 10/17/2018] [Accepted: 12/21/2018] [Indexed: 12/30/2022]
Abstract
Warfarin has a very narrow therapeutic window and obvious interindividual variability in its effects, with many factors contributing to the body's response. Algorithms incorporating multiple genetic, environment and clinical factors have been established to select a precision dose for each patient. A number of randomized controlled trials (RCTs) were conducted to explore whether patients could benefit from these algorithms; however, the results were inconsistent. Some questions remain to be resolved. Recently, new genetic and non-genetic factors have been discovered to contribute to variability in optimal warfarin doses. The results of further RCTs have been unveiled, and guidelines for pharmacogenetically guided warfarin dosing have been updated. Based on these most recent advancements, we summarize some open questions in this field and try to propose possible strategies to resolve them.
Collapse
Affiliation(s)
- Xi Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, P. R. China.,Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha, 410078, P. R. China
| | - Dan Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, P. R. China.,Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha, 410078, P. R. China
| | - Ji-Chu Wu
- Department of Cardiovascular, Central Hospital of Shaoyang, Shaoyang, 422000, P. R. China
| | - Zhao-Qian Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, P. R. China.,Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha, 410078, P. R. China
| | - Hong-Hao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, P. R. China.,Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha, 410078, P. R. China
| | - Ji-Ye Yin
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, P. R. China. .,Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha, 410078, P. R. China. .,Hunan Provincial Gynecological Cancer Diagnosis and Treatment Engineering Research Center, Changsha, 410078, P. R. China.
| |
Collapse
|
131
|
Friedman PN, Shaazuddin M, Gong L, Grossman RL, Harralson AF, Klein TE, Lee NH, Miller DC, Nutescu EA, O'Brien TJ, O'Donnell PH, O'Leary KJ, Tuck M, Meltzer DO, Perera MA. The ACCOuNT Consortium: A Model for the Discovery, Translation, and Implementation of Precision Medicine in African Americans. Clin Transl Sci 2019; 12:209-217. [PMID: 30592548 PMCID: PMC6510376 DOI: 10.1111/cts.12608] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 11/26/2018] [Indexed: 01/06/2023] Open
Abstract
The majority of pharmacogenomic (PGx) studies have been conducted on European ancestry populations, thereby excluding minority populations and impeding the discovery and translation of African American–specific genetic variation into precision medicine. Without accounting for variants found in African Americans, clinical recommendations based solely on genetic biomarkers found in European populations could result in misclassification of drug response in African American patients. To address these challenges, we formed the Transdisciplinary Collaborative Center (TCC), African American Cardiovascular Pharmacogenetic Consortium (ACCOuNT), to discover novel genetic variants in African Americans related to clinically actionable cardiovascular phenotypes and to incorporate African American–specific sequence variations into clinical recommendations at the point of care. The TCC consists of two research projects focused on discovery and translation of genetic findings and four cores that support the projects. In addition, the largest repository of PGx information on African Americans is being established as well as lasting infrastructure that can be utilized to spur continued research in this understudied population.
Collapse
Affiliation(s)
- Paula N Friedman
- Department of Pharmacology, Center for Pharmacogenomics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Mohammed Shaazuddin
- Department of Pharmacology, Center for Pharmacogenomics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Li Gong
- Department of Biomedical Data Science, Stanford University, Stanford, California, USA
| | - Robert L Grossman
- Department of Medicine and Computer Science, Center for Data Intensive Science, The University of Chicago, Chicago, Illinois, USA
| | - Arthur F Harralson
- Department of Pharmacogenomics, Shenandoah University, Inova Center for Personalized Health, Fairfax, Virginia, USA.,Department of Pharmacology and Physiology, The George Washington University, School of Medicine and Health Sciences, Washington, DC, USA
| | - Teri E Klein
- Department of Biomedical Data Science and Department of Medicine, Stanford University, Stanford, California, USA
| | - Norman H Lee
- Department of Pharmacology and Physiology, The George Washington University, School of Medicine and Health Sciences, Washington, DC, USA
| | - Doriane C Miller
- Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Edith A Nutescu
- Department of Pharmacy Systems, Outcomes, and Policy and Personalized Medicine Program, University of Illinois, College of Pharmacy, Chicago, Illinois, USA
| | - Travis J O'Brien
- Department of Pharmacology and Physiology, The George Washington University, School of Medicine and Health Sciences, Washington, DC, USA
| | - Peter H O'Donnell
- Department of Medicine, Center for Personalized Therapeutics, Committee on Clinical Pharmacology and Pharmacogenomics, The University of Chicago, Chicago, Illinois, USA
| | - Kevin J O'Leary
- Division of Hospital Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Matthew Tuck
- Washington DC VA Medical Center, Washington, DC and The George Washington University, Washington, DC, USA
| | - David O Meltzer
- Department of Medicine, Center for Health and the Social Sciences, The University of Chicago, Chicago, Illinois, USA
| | - Minoli A Perera
- Department of Pharmacology, Center for Pharmacogenomics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
132
|
International survey of patients undergoing percutaneous coronary intervention and their attitudes toward pharmacogenetic testing. Pharmacogenet Genomics 2019; 29:76-83. [PMID: 30724853 DOI: 10.1097/fpc.0000000000000368] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To evaluate perceptions toward pharmacogenetic testing of patients undergoing percutaneous coronary intervention (PCI) who are prescribed dual antiplatelet therapy (DAPT) and whether geographical differences in these perceptions exist. PARTICIPANTS AND METHODS TAILOR-PCI is the largest genotype-based cardiovascular clinical trial randomizing participants to conventional DAPT or prospective genotyping-guided DAPT. Enrolled patients completed surveys before and 6 months after randomization. RESULTS A total of 1327 patients completed baseline surveys of whom 28, 29, and 43% were from Korea, Canada and the USA, respectively. Most patients (77%) valued identifying pharmacogenetic variants; however, fewer Koreans (44%) as compared with Canadians (91%) and USA (89%) patients identified pharmacogenetics as being important (P<0.001). After adjusting for age, sex, and country, those who were confident in their ability to understand genetic information were significantly more likely to value identifying pharmacogenetic variants (odds ratio: 30.0; 95% confidence interval: 20.5-43.8). Only 21% of Koreans, as opposed to 86 and 77% of patients in Canada and USA, respectively, were confident in their ability to understand genetic information (P<0.001). CONCLUSION Although genetically mediated clopidogrel resistance is more prevalent amongst Asians, Koreans undergoing PCI identified pharmacogenetic variants as less important to their healthcare, likely related to their lack of confidence in their ability to understand genetic information. To enable successful implementation of pharmacogenetic testing on a global scale, the possibility of international population differences in perceptions should be considered.
Collapse
|
133
|
Vassy JL, Stone A, Callaghan JT, Mendes M, Meyer LJ, Pratt VM, Przygodzki RM, Scheuner MT, Wang-Rodriguez J, Schichman SA. Pharmacogenetic testing in the Veterans Health Administration (VHA): policy recommendations from the VHA Clinical Pharmacogenetics Subcommittee. Genet Med 2019; 21:382-390. [PMID: 29858578 PMCID: PMC6274593 DOI: 10.1038/s41436-018-0057-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 04/26/2018] [Indexed: 11/18/2022] Open
Abstract
PURPOSE The Veterans Health Administration (VHA) Clinical Pharmacogenetics Subcommittee is charged with making recommendations about whether specific pharmacogenetic tests should be used in healthcare at VHA facilities. We describe a process to inform VHA pharmacogenetic testing policy. METHODS After developing consensus definitions of clinical validity and utility, the Subcommittee identified salient drug-gene pairs with potential clinical application in VHA. Members met monthly to discuss each drug-gene pair, the evidence of clinical utility for the associated pharmacogenetic test, and any VHA-specific testing considerations. The Subcommittee classified each test as strongly recommended, recommended, or not routinely recommended before drug initiation. RESULTS Of 30 drug-gene pair tests reviewed, the Subcommittee classified 4 (13%) as strongly recommended, including HLA-B*15:02 for carbamazepine-associated Stevens-Johnston syndrome and G6PD for rasburicase-associated hemolytic anemia; 12 (40%) as recommended, including CYP2D6 for codeine toxicity; and 14 (47%) as not routinely recommended, such as CYP2C19 for clopidogrel dosing. CONCLUSION Only half of drug-gene pairs with high clinical validity received Subcommittee support for policy promoting their widespread use across VHA. The Subcommittee generally found insufficient evidence of clinical utility or available, effective alternative strategies for the remainders. Continual evidence review and rigorous outcomes research will help promote the translation of pharmacogenetic discovery to healthcare.
Collapse
Affiliation(s)
- Jason L Vassy
- Section of General Internal Medicine, VA Boston Healthcare System, Boston, Massachusetts, USA.
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA.
- Division of General Internal Medicine and Primary Care, Brigham and Women's Hospital, Boston, Massachusetts, USA.
| | - Annjanette Stone
- Pharmacogenomics Analysis Laboratory, Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA
| | - John T Callaghan
- Richard L. Roudebush VA Medical Center, Indianapolis, Indiana, USA
- Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | - Laurence J Meyer
- Office of Specialty Care Services, Veterans Health Administration, Washington, DC, USA
| | - Victoria M Pratt
- Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ronald M Przygodzki
- Office of Research & Development, US Department of Veterans Affairs, Washington, DC, USA
| | - Maren T Scheuner
- Division of Medical Genetics, Department of Medicine, VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
- Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, USA
| | - Jessica Wang-Rodriguez
- VA San Diego Healthcare System, San Diego, California, USA
- University of California San Diego, San Diego, California, USA
| | - Steven A Schichman
- Pharmacogenomics Analysis Laboratory, Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA
| |
Collapse
|
134
|
Schwarz UI, Gulilat M, Kim RB. The Role of Next-Generation Sequencing in Pharmacogenetics and Pharmacogenomics. Cold Spring Harb Perspect Med 2019; 9:cshperspect.a033027. [PMID: 29844222 DOI: 10.1101/cshperspect.a033027] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Inherited genetic variations in pharmacogenetic loci are widely acknowledged as important determinants of phenotypic differences in drug response, and may be actionable in the clinic. However, recent studies suggest that a considerable number of novel rare variants in pharmacogenes likely contribute to a still unexplained fraction of the observed interindividual variability. Next-generation sequencing (NGS) represents a rapid, relatively inexpensive, large-scale DNA sequencing technology with potential relevance as a comprehensive pharmacogenetic genotyping platform to identify genetic variation related to drug therapy. However, many obstacles remain before the clinical use of NGS-based test results, including technical challenges, functional interpretation, and strict requirements for diagnostic tests. Advanced computational analyses, high-throughput screening methodologies, and generation of shared resources with cell-based and clinical information will facilitate the integration of NGS data into candidate genotyping approaches, likely enhancing future drug phenotype predictions in patients.
Collapse
Affiliation(s)
- Ute I Schwarz
- Division of Clinical Pharmacology, Department of Medicine, Western University, London, Ontario N6A 5A5, Canada.,Department of Physiology and Pharmacology, Western University, London, Ontario N6A 5A5, Canada
| | - Markus Gulilat
- Department of Physiology and Pharmacology, Western University, London, Ontario N6A 5A5, Canada
| | - Richard B Kim
- Division of Clinical Pharmacology, Department of Medicine, Western University, London, Ontario N6A 5A5, Canada.,Department of Physiology and Pharmacology, Western University, London, Ontario N6A 5A5, Canada
| |
Collapse
|
135
|
Scott ER, Wallsten RL. A Look to the Future. Pharmacogenomics 2019. [DOI: 10.1016/b978-0-12-812626-4.00010-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
136
|
|
137
|
Yang T, Zhou Y, Chen C, Lu M, Ma L, Cui Y. Genotype-guided dosing versus conventional dosing of warfarin: A meta-analysis of 15 randomized controlled trials. J Clin Pharm Ther 2018; 44:197-208. [PMID: 30593674 DOI: 10.1111/jcpt.12782] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/31/2018] [Accepted: 11/18/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Ting Yang
- Department of Pharmacy; Peking University First Hospital; Beijing China
| | - Ying Zhou
- Department of Pharmacy; Peking University First Hospital; Beijing China
| | - Chaoyang Chen
- Department of Pharmacy; Peking University First Hospital; Beijing China
| | - Min Lu
- Department of Pharmacy; Peking University First Hospital; Beijing China
| | - Lingyue Ma
- Department of Pharmacy; Peking University First Hospital; Beijing China
| | - Yimin Cui
- Department of Pharmacy; Peking University First Hospital; Beijing China
| |
Collapse
|
138
|
Zhou S, Skaar DJ, Jacobson PA, Huang RS. Pharmacogenomics of Medications Commonly Used in the Intensive Care Unit. Front Pharmacol 2018; 9:1436. [PMID: 30564130 PMCID: PMC6289166 DOI: 10.3389/fphar.2018.01436] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 11/20/2018] [Indexed: 12/11/2022] Open
Abstract
In the intensive care unit (ICU) setting, where highly variable and insufficient drug efficacies, as well as frequent and unpredictable adverse drug reactions (ADRs) occur, pharmacogenomics (PGx) offers an opportunity to improve health outcomes. However, PGx has not been fully evaluated in the ICU, partly due to lack of knowledge of how genetic markers may affect drug therapy. To fill in this gap, we conducted a review to summarize the PGx information for the medications commonly encountered in the ICU.
Collapse
Affiliation(s)
- Shuqin Zhou
- Department of Emergency and Critical Care Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China.,Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, United States
| | - Debra J Skaar
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, United States
| | - Pamala A Jacobson
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, United States
| | - R Stephanie Huang
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
139
|
Xu Z, Zhang SY, Huang M, Hu R, Li JL, Cen HJ, Wang ZP, Ou JS, Yin SL, Xu YQ, Wu ZK, Zhang X. Genotype-Guided Warfarin Dosing in Patients With Mechanical Valves: A Randomized Controlled Trial. Ann Thorac Surg 2018; 106:1774-1781. [DOI: 10.1016/j.athoracsur.2018.07.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 06/30/2018] [Accepted: 07/06/2018] [Indexed: 12/27/2022]
|
140
|
Shendre A, Dillon C, Limdi NA. Pharmacogenetics of warfarin dosing in patients of African and European ancestry. Pharmacogenomics 2018; 19:1357-1371. [PMID: 30345882 PMCID: PMC6562764 DOI: 10.2217/pgs-2018-0146] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 09/28/2018] [Indexed: 12/20/2022] Open
Abstract
Despite the introduction of direct acting oral anticoagulants, warfarin remains the most commonly prescribed oral anticoagulant. However, warfarin therapy is plagued by the large inter- and intrapatient variability. The variability in dosing fueled research to identify clinical and genetic predictors and develop more accurate dosing algorithms. Observational studies have demonstrated the significant impact of single nucleotide polymorphisms in CYP2C9 and VKORC1 on warfarin dose in patients of European ancestry and African-Americans. This evidence supported the design and conduct of clinical trials to assess whether genotype-guided dosing results in improved anticoagulation control and outcomes. The trial results have shown discordance by race, with pharmacogenetic algorithms improving dose and anticoagulation control among European ancestry patients compared with African-American patients. Herein, we review the evidence from observational and interventional studies, highlight the need for inclusion of minority race groups and propose the need to develop race specific dosing algorithms.
Collapse
Affiliation(s)
- Aditi Shendre
- Department of Epidemiology, Richard M Fairbanks School of Public Health, Indiana University Purdue University Indianapolis, IN 46202, USA
| | - Chrisly Dillon
- Department of Neurology, School of Medicine, University of Alabama at Birmingham, AL 35294, USA
| | - Nita A Limdi
- Department of Neurology, School of Medicine, University of Alabama at Birmingham, AL 35294, USA
| |
Collapse
|
141
|
Effect of genetic and patient factors on warfarin pharmacodynamics following warfarin withdrawal: Implications for patients undergoing surgery. Thromb Res 2018; 171:167-170. [DOI: 10.1016/j.thromres.2018.09.064] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/27/2018] [Accepted: 09/28/2018] [Indexed: 01/18/2023]
|
142
|
Ma Z, Wang P, Gao Z, Wang R, Khalighi K. Ensemble of machine learning algorithms using the stacked generalization approach to estimate the warfarin dose. PLoS One 2018; 13:e0205872. [PMID: 30339708 PMCID: PMC6195267 DOI: 10.1371/journal.pone.0205872] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 10/02/2018] [Indexed: 11/19/2022] Open
Abstract
Warfarin dosing remains challenging due to narrow therapeutic index and highly individual variability. Incorrect warfarin dosing is associated with devastating adverse events. Remarkable efforts have been made to develop the machine learning based warfarin dosing algorithms incorporating clinical factors and genetic variants such as polymorphisms in CYP2C9 and VKORC1. The most widely validated pharmacogenetic algorithm is the IWPC algorithm based on multivariate linear regression (MLR). However, with only a single algorithm, the prediction performance may reach an upper limit even with optimal parameters. Here, we present novel algorithms using stacked generalization frameworks to estimate the warfarin dose, within which different types of machine learning algorithms function together through a meta-machine learning model to maximize the prediction accuracy. Compared to the IWPC-derived MLR algorithm, Stack 1 and 2 based on stacked generalization frameworks performed significantly better overall. Subgroup analysis revealed that the mean of the percentage of patients whose predicted dose of warfarin within 20% of the actual stable therapeutic dose (mean percentage within 20%) for Stack 1 was improved by 12.7% (from 42.47% to 47.86%) in Asians and by 13.5% (from 22.08% to 25.05%) in the low-dose group compared to that for MLR, respectively. These data suggest that our algorithms would especially benefit patients requiring low warfarin maintenance dose, as subtle changes in warfarin dose could lead to adverse clinical events (thrombosis or bleeding) in patients with low dose. Our study offers novel pharmacogenetic algorithms for clinical trials and practice.
Collapse
Affiliation(s)
- Zhiyuan Ma
- Easton Cardiovascular Associates, Easton, PA, United States of America
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC, United States of America
| | - Ping Wang
- Easton Cardiovascular Associates, Easton, PA, United States of America
| | - Zehui Gao
- Department of Mathematics and Statistics, San Diego State University, La Mesa, CA, United States of America
| | - Ruobing Wang
- Department of Chemistry and Social Science Research Institute, Duke University, Durham, NC, United States of America
- Science Center of Opera Solutions LLC, San Diego, CA, United States of America
| | - Koroush Khalighi
- Easton Cardiovascular Associates, Easton, PA, United States of America
- Division of Cardiology, Department of Medicine, Easton Hospital, Easton, PA, United States of America
- Drexel University College of Medicine, Philadelphia, PA, United States of America
| |
Collapse
|
143
|
Abstract
Pharmacogenomics is a tool for practitioners to provide precision pharmacotherapy using genomics. All providers are likely to encounter genomic data in practice with the expectation that they are able to successfully apply it to patient care. Pharmacogenomics tests for genetic variations in genes that are responsible for drug metabolism, transport, and targets of drug action. Variations can increase the risk for drug toxicity or poor efficacy. Pharmacogenomics can, therefore, be used to help select the best medication or aid in dosing. Nephrologists routinely treat cardiovascular disease and manage patients after kidney transplantation, two situations for which there are several high-evidence clinical recommendations for commonly used anticoagulants, antiplatelets, statins, and transplant medications. Successful use of pharmacogenomics in practice requires that providers are familiar with how to access and use pharmacogenomics resources. Similarly, clinical decision making related to whether to use existing data, whether to order testing, and if data should be used in practice is needed to deliver precision medicine. Pharmacogenomics is applicable to virtually every medical specialty, and nephrologists are well positioned to be implementation leaders.
Collapse
Affiliation(s)
| | | | - Philip E. Empey
- Department of Pharmacy and Therapeutics, School of Pharmacy, and
- Institute and of Precision Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
144
|
De T, Park CS, Perera MA. Cardiovascular Pharmacogenomics: Does It Matter If You're Black or White? Annu Rev Pharmacol Toxicol 2018; 59:577-603. [PMID: 30296897 DOI: 10.1146/annurev-pharmtox-010818-021154] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Race and ancestry have long been associated with differential risk and outcomes to disease as well as responses to medications. These differences in drug response are multifactorial with some portion associated with genomic variation. The field of pharmacogenomics aims to predict drug response in patients prior to medication administration and to uncover the biological underpinnings of drug response. The field of human genetics has long recognized that genetic variation differs in frequency between ancestral populations, with some single nucleotide polymorphisms found solely in one population. Thus far, most pharmacogenomic studies have focused on individuals of European and East Asian ancestry, resulting in a substantial disparity in the clinical utility of genetic prediction for drug response in US minority populations. In this review, we discuss the genetic factors that underlie variability to drug response and known pharmacogenomic associations and how these differ between populations, with an emphasis on the current knowledge in cardiovascular pharmacogenomics.
Collapse
Affiliation(s)
- Tanima De
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA;
| | - C Sehwan Park
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA;
| | - Minoli A Perera
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA;
| |
Collapse
|
145
|
Tully PJ, Helmer C, Peters R, Tzourio C. Exploiting Drug-Apolipoprotein E Gene Interactions in Hypertension to Preserve Cognitive Function: The 3-City Cohort Study. J Am Med Dir Assoc 2018; 20:188-194.e4. [PMID: 30292766 DOI: 10.1016/j.jamda.2018.08.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 08/01/2018] [Accepted: 08/03/2018] [Indexed: 01/29/2023]
Abstract
OBJECTIVES The objective was to test the hypothesis that antihypertensive drugs have a differential effect on cognition in carriers and noncarriers of the apolipoprotein ε4 (APOE4) polymorphism. DESIGN Prospective population-based cohort, France. SETTING AND PARTICIPANTS A total of 3359 persons using antihypertensive drugs (median age 74 years, 62% women) were serially assessed up to 10 years follow-up. MEASURES Exposure to antihypertensive drug use was established in the first 2 years. Cognitive function was assessed at baseline, 2, 4, 7, and 10 years with a validated test battery covering global cognition, verbal fluency, immediate visual recognition memory, processing speed, and executive function. Clinically significant change in cognitive function was determined using reliable change indices represented as z scores and analyzed with linear mixed-models. RESULTS From 3359 persons exposed to antihypertensive drugs, 653 were APOE4 carriers (5.1% homozygous, 94.9% heterozygous) and median follow-up was 5.2 years (interquartile range 3.7-8.0). In APOE4 carriers, improved general cognitive function over time was associated with exposure to angiotensin converting enzyme inhibitors [β = .14; 95% confidence interval (CI) .06-.23, P = .001] and angiotensin receptor blockers (β = .11; 95% CI .02-.21, P = .019). Improved verbal fluency was associated with angiotensin converting enzyme inhibitors (β = .11; 95% CI .03-.20, P = .012). CONCLUSIONS Renin-angiotensin-system blockade was associated with improved general cognitive function in APOE4 carriers. Findings did not support renin-angiotensin-system drugs' lipophilicity or ability to cross the blood-brain barrier as potential mechanisms. The findings have implications for selecting the optimal antihypertensive drug in older populations at risk of cognitive decline and dementia.
Collapse
Affiliation(s)
- Phillip J Tully
- University of Bordeaux, INSERM, Bordeaux Population Health Research Center, Team HEALTHY, Bordeaux, France; Freemasons Foundation Centre for Men's Health, Discipline of Medicine, School of Medicine, The University of Adelaide, Adelaide, Australia.
| | - Catherine Helmer
- University of Bordeaux, INSERM, Bordeaux Population Health Research Center, Team LEHA, Bordeaux, France
| | - Ruth Peters
- Center for Research on Ageing, Health and Wellbeing, Research School of Population Health, The Australian National University, Canberra, ACT, Australia; Imperial Clinical Trials Unit, School of Public Health, Imperial College London, London, United Kingdom
| | - Christophe Tzourio
- University of Bordeaux, INSERM, Bordeaux Population Health Research Center, Team HEALTHY, Bordeaux, France
| |
Collapse
|
146
|
Luzum JA, Cheung JC. Does cardiology hold pharmacogenetics to an inconsistent standard? A comparison of evidence among recommendations. Pharmacogenomics 2018; 19:1203-1216. [PMID: 30196751 PMCID: PMC6219446 DOI: 10.2217/pgs-2018-0097] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 08/17/2018] [Indexed: 12/20/2022] Open
Abstract
Current guideline recommendations for pharmacogenetic testing for clopidogrel by the American Heart Association/American College of Cardiology (AHA/ACC) contradict the Clinical Pharmacogenetics Implementation Consortium and the US FDA. The AHA/ACC recommends against routine pharmacogenetic testing for clopidogrel because no randomized controlled trials have demonstrated that testing improves patients' outcomes. However the AHA/ACC and the National Comprehensive Cancer Network (NCCN) recommend other pharmacogenetic tests in the absence of randomized controlled trials evidence. Using clopidogrel as a case example, we compared the evidence for other pharmacogenetic tests recommended by the AHA/ACC and NCCN. In patients that received percutaneous coronary intervention, the evidence supporting pharmacogenetic testing for clopidogrel is stronger than other pharmacogenetic tests recommended by the AHA/ACC and NCCN.
Collapse
Affiliation(s)
- Jasmine A Luzum
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Jason C Cheung
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
- Department of Pharmacy, Baptist Health Floyd, New Albany, IN, USA
| |
Collapse
|
147
|
Palmer A, Chen A, Matsumoto T, Murphy M, Price A. Blood management in total knee arthroplasty: state-of-the-art review. J ISAKOS 2018. [DOI: 10.1136/jisakos-2017-000168] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Total blood loss from primary total knee arthroplasty may exceed 2 L with greater blood loss during revision procedures. Blood loss and allogeneic transfusion are strongly associated with adverse outcomes from surgery including postoperative mortality, thromboembolic events and infection. Strategies to reduce blood loss and transfusion rates improve patient outcomes and reduce healthcare costs. Interventions are employed preoperatively, intraoperatively and postoperatively. The strongest predictor for allogeneic blood transfusion is preoperative anaemia. Over 35% of patients are anaemic when scheduled for primary and revision knee arthroplasty, defined as haemoglobin <130 g/L for men and women, and the majority of cases are secondary to iron deficiency. Early identification and treatment of anaemia can reduce postoperative transfusions and complications. Anticoagulation must be carefully managed perioperatively to balance the risk of thromboembolic event versus the risk of haemorrhage. Intraoperatively, tranexamic acid reduces blood loss and is recommended for all knee arthroplasty surgery; however, the optimal route, dose or timing of administration remains uncertain. Cell salvage is a valuable adjunct to surgery with significant expected blood loss, such as revision knee arthroplasty. Autologous blood donation is not recommended in routine care, sealants may be beneficial in select cases but further evidence of benefit is required, and the use of a tourniquet remains at the discretion of the surgeon. Postoperatively, restrictive transfusion protocols should be followed with a transfusion threshold haemoglobin of 70 g/L, except in the presence of acute coronary syndrome. Recent studies report no allogeneic transfusions after primary knee arthroplasty surgery after employing blood conservation strategies. The current challenge is to select and integrate different blood conserving interventions to deliver an optimal patient pathway with a multidisciplinary approach.
Collapse
|
148
|
Wu AH. Promoting the value of precision medicine to the public: the power of storytelling. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2018. [DOI: 10.1080/23808993.2018.1506255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Alan. H.B Wu
- Department of Laboratory Medicine, University of California, San Francisco San Francisco General Hospital, San Francisco, CA, USA
| |
Collapse
|
149
|
Tse G, Gong M, Li G, Wong SH, Wu WKK, Wong WT, Roever L, Lee APW, Lip GYH, Wong MCS, Liu T. Genotype-guided warfarin dosing vs. conventional dosing strategies: a systematic review and meta-analysis of randomized controlled trials. Br J Clin Pharmacol 2018; 84:1868-1882. [PMID: 29704269 PMCID: PMC6089819 DOI: 10.1111/bcp.13621] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 04/03/2018] [Accepted: 04/17/2018] [Indexed: 12/20/2022] Open
Abstract
AIMS Previous trials on the effectiveness of genotype-guided warfarin dosing vs. conventional dosing have been inconclusive. We conducted a systematic review and meta-analysis of randomized trials comparing genotype-guided to conventional dosing strategies. METHODS PubMed and the Cochrane Library were searched up to 23 October 2017. RESULTS A total of 76 and 94 entries were retrieved were retrieved from PubMed and the Cochrane Library, respectively. A total of 2626 subjects in the genotype-guided dosing (mean age 63.3 ± 5.8 years; 46% male) and 2604 subjects in the conventional dosing (mean age 64.7 ± 6.1 years; 46% male) groups (mean follow-up duration 64 days) from 18 trials were included. Compared with conventional dosing, genotype-guided dosing significantly shortened the time to first therapeutic international normalized ratio (INR) (mean difference 2.6 days, standard error 0.3 days; P < 0.0001; I2 0%) and time to first stable INR (mean difference 5.9 days, standard error 2.0 days; P < 0.01; I2 94%). Genotype-guided dosing also increased the time in therapeutic range (mean difference 3.1%, standard error 1.2%; P < 0.01; I2 80%) and reduced the risks of both excessive anticoagulation, defined as INR ≥4 [risk ratio (RR) 0.87; 95% confidence interval (CI) 0.78, 0.98; P < 0.05; I2 : 0%), and bleeding (RR 0.82; 95% CI 0.69, 0.98; P < 0.05; I2 31%). No difference in thromboembolism (RR 0.84; 95% CI 0.56, 1.26; P = 0.40; I2 0%) or mortality (RR 1.16; 95% CI 0.46, 2.91; P = 0.76; I2 0%) was observed between the two groups. CONCLUSIONS Genotype-guided warfarin dosing offers better safety with less bleeding compared with conventional dosing strategies. No significant benefit on thromboembolism or mortality was evident.
Collapse
Affiliation(s)
- Gary Tse
- Department of Medicine and Therapeutics, Faculty of MedicineChinese University of Hong KongHong KongSARPeople's Republic of China
- Li Ka Shing Institute of Health Sciences, Faculty of MedicineChinese University of Hong KongHong KongSARPeople's Republic of China
| | - Mengqi Gong
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of CardiologySecond Hospital of Tianjin Medical UniversityTianjin300211People's Republic of China
| | - Guangping Li
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of CardiologySecond Hospital of Tianjin Medical UniversityTianjin300211People's Republic of China
| | - Sunny Hei Wong
- Department of Medicine and Therapeutics, Faculty of MedicineChinese University of Hong KongHong KongSARPeople's Republic of China
- Li Ka Shing Institute of Health Sciences, Faculty of MedicineChinese University of Hong KongHong KongSARPeople's Republic of China
| | - William K. K. Wu
- Li Ka Shing Institute of Health Sciences, Faculty of MedicineChinese University of Hong KongHong KongSARPeople's Republic of China
- Department of Anaesthesia and Intensive Care, State Key Laboratory of Digestive DiseaseThe Chinese University of Hong KongHong KongSARPeople's Republic of China
| | - Wing Tak Wong
- School of Life SciencesChinese University of Hong KongHong KongSARPeople's Republic of China
| | - Leonardo Roever
- Department of Clinical ResearchFederal University of UberlândiaMGBrazil
| | - Alex Pui Wai Lee
- Department of Medicine and Therapeutics, Faculty of MedicineChinese University of Hong KongHong KongSARPeople's Republic of China
| | - Gregory Y. H. Lip
- Institute of Cardiovascular SciencesUniversity of BirminghamBirminghamUK
- Aalborg Thrombosis Research Unit, Department of Clinical MedicineAalborg UniversityAalborgDenmark
| | - Martin C. S. Wong
- JC School of Public Health and Primary CareThe Chinese University of Hong KongHong KongSARPeople's Republic of China
- State Key Laboratory of Digestive DiseaseChinese University of Hong KongHong KongSARPeople's Republic of China
| | - Tong Liu
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of CardiologySecond Hospital of Tianjin Medical UniversityTianjin300211People's Republic of China
| | | |
Collapse
|
150
|
Tse G, Roever L, Wong MCS, Lip GYH, Liu T. Reply to 'Comment on 'Genotype-guided warfarin dosing vs. conventional dosing strategies: a systematic review and meta-analysis of randomized clinical trials' by Tse et al.'. Br J Clin Pharmacol 2018; 84:2165-2166. [PMID: 29984539 PMCID: PMC6089818 DOI: 10.1111/bcp.13658] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 05/23/2018] [Accepted: 05/29/2018] [Indexed: 01/13/2023] Open
Affiliation(s)
- Gary Tse
- Department of Medicine and TherapeuticsFaculty of Medicine, Chinese University of Hong KongHong KongSARChina
- Li Ka Shing Institute of Health SciencesFaculty of Medicine, Chinese University of Hong KongHong KongSARChina
| | - Leonardo Roever
- Department of Clinical ResearchFederal University of UberlândiaUberlândiaMGBrazil
| | - Martin C. S. Wong
- JC School of Public Health and Primary CareChinese University of Hong KongHong KongSARChina
- State Key Laboratory of Digestive DiseaseChinese University of Hong KongHong KongSARChina
| | - Gregory Y. H. Lip
- Institute of Cardiovascular SciencesUniversity of BirminghamBirminghamUK
- Aalborg Thrombosis Research Unit, Department of Clinical MedicineAalborg UniversityAalborgDenmark
| | - Tong Liu
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of CardiologyTianjin Institute of Cardiology, Second Hospital of Tianjin Medical UniversityTianjinChina
| | | |
Collapse
|